Language selection

Search

Patent 2843771 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2843771
(54) English Title: ANTI-C-MET ANTIBODIES
(54) French Title: ANTICORPS ANTI-C-MET
Status: Deemed Abandoned and Beyond the Period of Reinstatement - Pending Response to Notice of Disregarded Communication
Bibliographic Data
(51) International Patent Classification (IPC):
  • C7K 16/28 (2006.01)
  • C7K 16/30 (2006.01)
  • C12P 21/08 (2006.01)
  • G1N 33/577 (2006.01)
(72) Inventors :
  • DAVIES, JULIAN (United States of America)
  • LIU, LING (United States of America)
  • LU, JIRONG (United States of America)
(73) Owners :
  • ELI LILLY AND COMPANY
(71) Applicants :
  • ELI LILLY AND COMPANY (United States of America)
(74) Agent: GOWLING WLG (CANADA) LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2012-09-13
(87) Open to Public Inspection: 2013-03-28
Examination requested: 2014-01-30
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2012/055057
(87) International Publication Number: US2012055057
(85) National Entry: 2014-01-30

(30) Application Priority Data:
Application No. Country/Territory Date
61/536,698 (United States of America) 2011-09-20
61/537,677 (United States of America) 2011-09-22

Abstracts

English Abstract

Provided is a monoclonal antibody, or antigen-binding fragment thereof, that binds to c-Met. Such antibodies, or antigen-binding fragments thereof, are useful in in vivo, ex vivo or in vitro immunochemical and other imaging methods for detecting cell surface c-Met receptor levels for diagnostic, prognostic and predictive purposes, and for optimizing therapeutic regimens in patients harboring tumors in which c-Met is implicated in pathogenesis.


French Abstract

L'invention concerne un anticorps monoclonal, ou un fragment de liaison à un antigène de celui-ci, qui se lie à c-Met. De tels anticorps, ou fragments de liaison à un antigène de ceux-ci, sont utiles dans des procédés immunochimiques et autres procédés d'imagerie in vivo, ex vivo ou in vitro pour la détection des niveaux de récepteur c-Met de surface cellulaire à des fins de diagnostic, de pronostic et de prédiction, et pour l'optimisation de régimes thérapeutiques chez des patients présentant des tumeurs dans lesquelles c-Met est mis en jeu dans la pathogenèse.

Claims

Note: Claims are shown in the official language in which they were submitted.


33
We Claim:
1. An antibody, or an antigen-binding Fab fragment thereof, that specifically
binds to the extracellular domain (ECD) of mature human c-Met consisting of
amino
acids 25-932 of SEQ ID NO: 19, the antibody or the Fab fragment comprising:
i. light chain complementarity determining regions LCDR1,
LCDR2,
and LCDR3 consisting of the amino acid sequences SVSSSISSTNLH (SEQ ID NO:
1), GTSNLAS (SEQ ID NO: 2), and QQWSSYPYT (SEQ ID NO: 3), respectively;
and
ii. heavy chain complementarity determining regions HCDR1,
HCDR2, and HCDR3 consisting of the amino acid sequences GYTFTSRYIH (SEQ
ID NO: 4), WIYPVTGDTYYNEKFKG (SEQ ID NO: 5), and GGGMFYY (SEQ ID
NO: 6), respectively.
2. The antibody, or the Fab fragment, of claim 1 comprising:
i. a light chain variable region (LCVR) comprising the amino
acid
sequence as in SEQ ID NO: 7; and
ii. a heavy chain variable region (HCVR) comprising the amino
acid
sequence as in SEQ ID NO: 9.
3. The antibody of claim 1 or 2 comprising:
i. a light chain comprising the amino acid sequence as in SEQ ID
NO: 11; and
ii. a heavy chain comprising the amino acid sequence as in SEQ
ID NO: 13.
4. The antibody of claim 3 comprising:
i. two light chains; and

34
ii. two heavy chains, wherein each of the light chains consist
of
the amino acid sequence as in SEQ ID NO: 11 and each of the
heavy chains consist of the amino acid sequence as in SEQ ID
NO: 13.
5. The antibody or the Fab fragment of claim 1 or 2, further comprising a
detectable label.
6. The antibody of claim 4, further comprising a detectable label.
7. A composition comprising:
a. an antibody, or an antigen-binding Fab fragment thereof, that
specifically binds to the extracellular domain (ECD) of mature human c-Met
consisting of amino acids 25-932 of SEQ ID NO: 19, the antibody or the Fab
fragment comprising:
i. light chain complementarity determining regions LCDR1,
LCDR2, and LCDR3 consisting of the amino acid sequences SVSSSISSTNLH (SEQ
ID NO: 1), GTSNLAS (SEQ ID NO: 2), and QQWSSYPYT (SEQ ID NO: 3),
respectively; and
ii. heavy chain complementarity determining regions HCDR1,
HCDR2, and HCDR3 consisting of the amino acid sequences GYTFTSRYIH (SEQ
ID NO: 4), WIYPVTGDTYYNEKFKG (SEQ ID NO: 5), and GGGMFYY (SEQ ID
NO: 6), respectively, and
b. a diagnostically acceptable carrier, diluent, or excipient.
8. The composition of claim 7, wherein the antibody or the Fab fragment
comprises:
i. a light chain variable region (LCVR) comprising the amino
acid sequence as in SEQ ID NO: 7; and
ii. a heavy chain variable region (HCVR) comprising the
amino acid sequence as in SEQ ID NO: 9.

35
9. A composition comprising the antibody or the Fab fragment of claim 1 or 2
and a polypeptide comprising the ECD of mature human c-Met consisting of amino
acids 25-932 of SEQ ID NO: 19, wherein the antibody or the Fab fragment is
bound
to the polypeptide.
10. The composition of claim 9, wherein the antibody or the Fab fragment is
bound to the polypeptide comprising the ECD of mature human c-Met at an
epitope
within the amino acid sequence as in SEQ ID NO: 15 or 16.
11. A kit comprising a container comprising the antibody or Fab fragment of
claim 1 or 2.
12. The kit of claim 11 further comprising a container comprising a secondary
antibody that binds to the antibody or the Fab fragment; and, optionally,
instructions
for using the antibody or the Fab fragment, with or without the secondary
antibody, to
detect c-Met.
13. The kit of claim 12, wherein said secondary antibody is conjugated to an
enzyme.
14. The kit of claim 13, further comprising a container having a chromogenic
substrate of said enzyme.
15. The antibody or the Fab fragment of claim 1 or 2 for use in diagnosis,
prognosis or prediction of a cancer treatment response to an anti-c-Met
therapeutic
antibody or chemotherapeutic agent.
16. A method of detecting c-Met expressed by a human cell, the method
comprising:
(a) contacting said cell with an antibody, or an antigen-binding
Fab fragment thereof, that specifically binds to the ECD of mature human c-Met
consisting of amino acids 25-932 of SEQ ID NO: 19, the antibody or the Fab
fragment comprising:

36
i. light chain complementarity determining regions LCDR1,
LCDR2,
and LCDR3 comprising the amino acid sequences SVSSSISSTNLH (SEQ ID NO: 1),
GTSNLAS (SEQ ID NO: 2), and QQWSSYPYT (SEQ ID NO: 3), respectively; and
ii. heavy chain complementarity determining regions HCDR1,
HCDR2, and HCDR3 comprising the amino acid sequences GYTFTSRYIH (SEQ ID
NO: 4), WIYPVTGDTYYNEKFKG (SEQ ID NO: 5), and GGGMFYY (SEQ ID NO:
6), respectively, for a time and under conditions that permit said antibody or
the Fab
fragment to bind to the ECD;
(b) optionally, removing any non-specifically bound antibody
or Fab fragment; and
(c) detecting and/or quantifying the amount of the antibody or the Fab
fragment which is specifically bound to the ECD.
17. A method of selecting a patient having a tumor suitable for treatment with
an anti-c-Met therapeutic antibody or chemotherapeutic agent, comprising:
(a) contacting at least one cell from said tumor with an antibody, or an
antigen-binding Fab fragment thereof, that specifically binds to the ECD of
mature
human c-Met consisting of amino acids 25-932 of SEQ ID NO: 19, the antibody or
the Fab fragment comprising:
i. light chain complementarity determining regions LCDR1,
LCDR2, and LCDR3 comprising the amino acid sequences SVSSSISSTNLH
(SEQ ID NO: 1), GTSNLAS (SEQ ID NO: 2), and QQWSSYPYT (SEQ ID
NO: 3), respectively; and
ii. heavy chain complementarity determining regions HCDR1,
HCDR2, and HCDR3 comprising the amino acid sequences GYTFTSRYIH (SEQ ID
NO: 4), WIYPVTGDTYYNEKFKG (SEQ ID NO: 5), and GGGMFYY (SEQ ID NO:
6), respectively, for a time and under conditions that permit said antibody,
or the Fab
fragment, to bind to the ECD;
(b) optionally, removing any non-specifically bound antibody or Fab
fragment; and
(c) detecting and/or quantifying the amount of antibody or

37
Fab fragment which is specifically bound to the ECD, wherein the presence of
the
antibody or Fab fragment specifically bound to the ECD identifies the patient
as being
appropriate for treatment with said anti-c-Met therapeutic antibody or
chemotherapeutic agent.
18. The method of claims 16 or 17, wherein said cell is an isolated cell, or
is
present in a cell pellet, a xenograft sample, a tissue sample, an organ
sample, or a
bodily fluid sample.
19. Use of a kit comprising the antibody or Fab fragment of claim 1 or 2 for:
(a) detecting and/or quantifying c-Met in or on a cell;
(b) detecting and/or quantifying c-Met expressing tumor cells in a
patient;
(c) detecting and/or quantifying c-Met expressing circulating tumor
cells in a blood sample of a patient;
(d) detecting and/or quantifying c-Met expressing tumor cells in a
bodily fluid from a cancer patient such as in blood, blood serum, urine,
ascites fluid,
lymphatic fluid, spinal fluid, and bronchial fluid;
(e) assessing whether an individual has, or is at risk for developing,
cancer of a tissue or organ wherein c-Met is expressed or over-expressed;
(f) selecting a patient having a tumor suitable for treatment with an
anti-c-Met therapeutic antibody or chemotherapeutic agent; or
(g) determining response to treatment with an anti-c-Met therapeutic
antibody or chemotherapeutic agent.
20. A method of predicting a patient's response to the administration of an
anti-c-Met therapeutic antibody or chemotherapeutic agent, comprising:
(a) contacting at least one cell from said patient's tumor with the
antibody or Fab fragment of claim 1 or 2 for a time and under conditions that
permit
said antibody or the Fab fragment to bind to the ECD of mature human c-Met;
(b) optionally, removing any non-specifically bound antibody or Fab
fragment; and
(c) detecting and/or quantifying the amount of the antibody or Fab

38
fragment which is specifically bound to the ECD of mature human c-Met, wherein
a
patient's response to the administration of the therapeutic antibody or
chemotherapeutic agent is predicted based on the presence of the antibody or
Fab
fragment of claim 1 specifically bound to the ECD of mature human c-Met.
21. A method of measuring a patient's response to the administration of an
anti-c-Met therapeutic antibody or chemotherapeutic agent, comprising:
(a) contacting at least one cell from a patient's tumor with the
antibody or the Fab fragment of claim 1 or claim 2 both (i) prior to and (ii)
after administration of an anti-c-Met therapeutic antibody or chemotherapeutic
agent to the patient for a time and under conditions that permit the antibody
or
the Fab fragment to bind to the ECD of mature human c-Met;
(b) optionally, removing any non-specifically bound antibody or Fab
fragment; and
(c) detecting and/or quantifying the amount of the antibody or Fab
fragment of claim 1 which is specifically bound to the ECD, wherein a response
to the
administration of the therapeutic antibody or chemotherapeutic agent to an
individual
is measured when the amount of the antibody or Fab fragment detected or
quantified
in (c) is greater in (i) as compared to (ii).

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
1
Anti-c-Met Antibodies
The present invention relates to the field of medicine. More particularly, the
invention relates to antibodies that bind c-Met to form a detectable c-
Met/anti-c-Met
monoclonal antibody complex useful in diagnostic techniques that require
labeling,
marking, or identifying c-Met such as imaging, prognostic or predictive
applications that
aid in identifying patients with tumors expressing high levels of c-Met and/or
improving
their treatment response with anti-c-Met therapeutics.
The protein c-Met is a member of the receptor tyrosine kinase superfamily, and
the receptor for hepatocyte growth factor (FIGF) also known as scatter factor
(SF). The
mature c-Met protein is composed of a completely extracellular alpha subunit,
a beta
subunit comprised of an extracellular ligand binding domain, a single
transmembrane
domain, and a cytoplasmic tyrosine kinase domain.
Activation of c-Met by HGF has been shown to enhance characteristics that are
associated with invasive cell phenotype: proliferation, migration,
morphogenesis,
survival (including protection from apoptosis), and protease synthesis. The c-
Met
signaling pathway is one of the most frequently dysregulated pathways in human
cancers,
and occurs in virtually all types of solid tumors (Knudsen et al. (2008)
Current Opinion in
Genetics & Development 18:87-96). The stimulation, overexpression, or mutation
of
c-Met is observed in many types of cancers, including colon, breast, ovary,
lung, liver,
prostate, thyroid, kidney, as well as melanomas and sarcomas. These
biochemical and
genetic abnormalities of the HGF/c-Met signaling axis are correlated with poor
clinical
outcomes and drug resistance in cancer patients (Liu, et al., Expert Opin.
lnvestig. Drugs,
17(7):997-1011 (2008)).
Due to the role of the c-Met signaling pathway in regulating initial steps of
tumor
formation and subsequent disease dissemination, c-Met is considered to be an
attractive
target for cancer therapy with small molecule and antibody antagonists of HGF
and/or
c-Met in development.
Prat, et al. (Mol. Cell. Biol.,11:5954-5962 (1991)) and PCT International
Publication WO 92/20792 disclose several monoclonal antibodies that bind the
extracellular domain (ECD) of the 0-chain of c-Met, and their use to detect
distribution of
c-Met in normal and neoplastic human tissues.

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
2
PCT International Publication WO 2009/029591 discloses a monoclonal antibody,
designated MET4, capable of staining c-Met in formalin fixed and paraffin
embedded
(FFPE) tumor tissues.
For evaluation of the level of expression of c-Met by tumor cells of a cancer
patient and/or determining if the treatment of a patient with one or more
therapeutic anti-
c-Met agents has resulted in a decreased level of c-Met expression by tumor
cells and/or a
reduced number of c-Met expressing tumor cells, there is a critical need for
c-Met diagnostic antibodies that can specifically bind to membrane localized c-
Met
particularly when the c-Met is already bound to a therapeutic anti-c-Met
agent. Dual
binding of a c-Met antibody of the present invention with an anti-c-Met
therapeutic agent
permits diagnostic assessment of tumor tissue, both before and after treatment
of the
patient, without compromising the desired diagnostic detection effect.
Accordingly, the invention provides alternative anti-c-Met antibodies that
specifically bind to the extracellular domain (ECD) of human c-Met. The anti-c-
Met
antibodies of the present invention may be useful as diagnostics to aid in
identification of
cancer patients with tumor cells expressing relatively high levels of c-Met.
Furthermore,
such anti-c-Met antibodies may be used to monitor and/or optimize a cancer
patient's
treatment with c-Met targeted therapeutic agents, such as the small molecule
antagonists
of c-Met described in WO 2010/011538 and U.S. Patent Application Publication
No. 2012/0028984 as well as antibody antagonists of c-Met such as those
described in
WO 2010/059654 and/or U.S. Patent No. 8,217,148.
One aspect of this invention pertains to a monoclonal antibody, or antigen-
binding
fragment thereof, that specifically binds to the ECD of human c-Met consisting
of the
amino acid sequence as in SEQ ID NO: 19, said antibody, or antigen-binding
fragment
thereof, comprising:
light chain complementarity determining regions LCDR1, LCDR2, and LCDR3
comprising the amino acid sequences SVSSSISSTNLH (SEQ ID NO: 1), GTSNLAS
(SEQ ID NO: 2), and QQWSSYPYT (SEQ ID NO: 3), respectively, and
heavy chain complementarity determining regions HCDR1, HCDR2, and HCDR3
comprising the amino acid sequences GYTFTSRYIH (SEQ ID NO: 4),
WIYPVTGDTYYNEKFKG (SEQ ID NO: 5), and GGGMFYY (SEQ ID NO: 6),
respectively.

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
3
Another embodiment of the invention is a monoclonal antibody, or antigen-
binding fragment thereof, that specifically binds to the ECD of human c-Met
consisting of
the amino acid sequence as in SEQ ID NO: 19, said monoclonal antibody
comprising:
a light chain variable region (LCVR) comprising the amino acid sequence as in
SEQ ID NO: 7, and
a heavy chain variable region (HCVR) comprises the amino acid sequence as in
SEQ ID NO: 9.
In another embodiment, the invention provides a monoclonal antibody, or
antigen-
binding fragment thereof, that specifically binds to the ECD of human c-Met
consisting of
the amino acid sequence as in SEQ ID NO: 19, said monoclonal antibody
comprising:
a light chain comprising the amino acid sequence as in SEQ ID NO: 11, and
a heavy chain comprising the amino acid sequence as in SEQ ID NO: 13.
In another embodiment, the invention provides a monoclonal antibody, or
antigen-
binding fragment thereof, that specifically binds to the ECD of human c-Met
consisting of
the amino acid sequence as in SEQ ID NO: 19, said monoclonal antibody
comprising two
light chains and two heavy chains, wherein each light chain is the polypeptide
of SEQ ID
NO: 11, and each heavy chain is the polypeptide of SEQ ID NO: 13.
Another embodiment of the invention provides a diagnostically useful
composition comprising one of the foregoing anti-c-Met monoclonal antibodies,
or
antigen-binding fragments thereof, of the present invention and a
diagnostically
acceptable carrier, diluent, or excipient. In one such embodiment, the c-Met
monoclonal
antibody, or antigen-binding fragment thereof, is covalently, non-covalently,
or partially
covalently and partially non-covalently linked to a detectable moiety.
In another embodiment, the invention encompasses a composition comprising at
least one of the foregoing anti-c-Met monoclonal antibodies, or antigen-
binding
fragments thereof, of the present invention bound to a polypeptide comprising
the ECD of
human c-Met consisting of the amino acid sequence as in SEQ ID NO: 19.
Preferably, an
anti-c-Met monoclonal antibody, or antigen-binding fragment thereof, of the
present
invention is bound to a polypeptide comprising the ECD of human c-Met
consisting of
the amino acid sequence as in SEQ ID NO: 19 at an epitope within the amino
acid
sequence as in SEQ ID NO: 15 or 16. More preferably, a composition comprising
at least
one of the foregoing anti-c-Met monoclonal antibodies, or antigen-binding
fragments

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
4
thereof, of the present invention bound to a polypeptide comprising the ECD of
human
c-Met consisting of the amino acid sequence as in SEQ ID NO: 19 further
comprises a
diagnostically acceptable carrier, diluent, or excipient.
In other embodiments, the invention provides a kit, comprising a container
comprising a monoclonal antibody, or antigen-binding fragment thereof, that
specifically
binds to the ECD of human c-Met consisting of the amino acid sequence as in
SEQ ID
NO: 19, said monoclonal antibody, or antigen-binding fragment thereof,
comprising:
light chain complementarity determining regions LCDR1, LCDR2, and LCDR3
comprising the amino acid sequences SVSSSISSTNLH (SEQ ID NO: 1), GTSNLAS
(SEQ. ID NO: 2), and QQWSSYPYT (SEQ. ID NO: 3), respectively, and
heavy chain complementarity determining regions HCDR1, HCDR2, and HCDR.3
comprising the amino acid sequences GYTFTSRYIH (SEQ ID NO: 4),
WIYPVTGDTYYNEKFKG (SEQ ID NO: 5), and GGGMFYY (SEQ ID NO: 6),
respectively. In certain embodiments, th.e kit may comprise a second container
comprising a secondary antibody that binds to the monoclonal antibody, or
antigen-
binding fragment thereof, and, optionally, instructions for using the
monoclonal antibody,
or antigen-binding fragment thereof, with or without the secondary antibody,
to detect
c-Met in vivo, ex vivo or in vitro. In some embodiments, the secondary
antibody can be
conjugated to an enzyme known to be used in immunoassays. In other
embodiments, the
kit can further comprise another container comprising a chromogenic substrate
of the
aforementioned enzyme. Preferably, the methods described herein are for
detecting c-
Met expression or over-expression are in vitro methods. Likewise, the kits
described
herein are used to detect c-Met expression or over-expression in vitro,
preferably.
In other embodiments, the invention also encompasses a method of detecting
c-Met expressed or overexpressed by a mammalian cell comprising: (a)
contacting a cell
with a detectably labeled monoclonal antibody, or antigen-binding fragment
thereof, that
specifically binds to the ECD of human c-Met consisting of the amino acid
sequence as in
SEQ ID NO: 19, said monoclonal antibody, or antigen-binding fragment thereof,
comprising:
light chain complementarity determining regions LCDR1, LCDR2, and LCDR3
comprising the amino acid sequences SVSSSISSTNLH (SEQ ID NO: 1), GTSNLAS
(SEQ ID NO: 2), and QQWSSYPYT (SEQ ID NO: 3), respectively, and

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
heavy chain complementarity determining regions HCDR1, HCDR2, and HCDR3
comprising the amino acid sequences GYTFTSRYIH (SEQ ID NO: 4),
WIYPVTGDTYYNEKFKG (SEQ ID NO: 5), and GGGMFYY (SEQ ID NO: 6), for a
time and under conditions that permit said monoclonal antibody to bind to the
ECD of the
5 c-Met expressed by the cell; (h) optionally, removing any non-
specifically bound
monoclonal antibody or antigen-binding fragment thereof; and (c) detecting
and/or
quantifying the amount of labeled monoclonal antibody or antigen-binding
fragment
thereof which is specifically bound to the ECD using any art-known method (for
example, cytometric techniques). Preferably, the c-Met is detected in a
biological tissue
or a bodily fluid obtained from a cancer patient such as in urine, ascites
fluid, lymphatic
fluid, spinal fluid, bronchial fluid, blood serum or, preferably, blood.
The invention also provides a method of detecting and/or quantifying tumor
cells
and/or circulating tumor cells expressing or over-expressing c-Met in a
mammalian
biological tissue or a bodily fluid obtained from a cancer patient such as in
urine, ascites
fluid, lymphatic fluid, spinal fluid, bronchial fluid, blood serum, or,
preferably, blood,
comprising: a step of contacting the tissue with a monoclonal antibody, or
antigen-
binding fragment thereof, that specifically binds to the ECD of human c-Met
consisting of
the amino acid sequence as in SEQ ID NO: 19, said monoclonal antibody, or
antigen-
binding fragment thereof, comprising:
light chain complementarity determining regions LCDR1, LCDR2, and LCDR3
comprising the amino acid sequences SVSSSISSTNLH (SEQ ID NO: 1), GTSNLAS
(SEQ ID NO: 2), and QQWSSYPYT (SEQ ID NO: 3), respectively, and
heavy chain complementarity determining regions HCDR1, HCDR2, and HCDR3
comprising the amino acid sequences GYTFTSRYIII (SEQ ID NO: 4),
WIYPVTGDTYYNEKFKG (SEQ ID NO: 5), and GGGMFYY (SEQ ID NO: 6) under
conditions that permit formation of a detectable c-Met/anti-c-Met monoclonal
antibody
complex; and a next step of detecting and/or quantifying said complex(es). In
various
embodiments, the contacting can be carried out in vitro. In various
embodiments, the
contacting can be carried out in vivo. For example, in various embodiments,
the anti-c-
Met monoclonal antibody, or antigen-binding fragment thereof, can be
administered
parenterally, by injection or infusion. Additionally, in various embodiments,
the anti-c-
Met monoclonal antibody, or antigen-binding fragment thereof, is labeled with
at least

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
6
one detectable agent including, but not limited to, the aforementioned labels.
Detection
can be performed in vitro or in vivo using a method, technology and/or device
lcnown in
the art to be appropriate for a specific detectable label including, but not
limited to, a
gamma counter, a scintillation counter, by autoradiography, and/or devices
including, for
example, a fluorescence measuring device, a bioluminescence measuring device,
a
magnetic resonance imaging (MRI) device, a magnetic device, a positron
emission
tomography (PET) device, a computed tomography (CT) device, an ultrasound
device, an
optical coherence tomography (OCT) device, and/or a single photon emission
computed
tomography (SPECT) device.
In another embodiment, the invention provides the use of a kit comprising a
monoclonal antibody, or antigen-binding fragment thereof, that specifically
binds to the
LCD of human c-Met consisting of the amino acid sequence as in SEQ ID NO: 19,
said
monoclonal antibody, or antigen-binding fragment thereof, comprising:
light chain complementarity determining regions LCDR.1, LCDR2, and LCDR3
comprising the amino acid sequences SVSSSISSTNLH (SEQ ID NO: 1), GTSNLAS
(SEQ ID NO: 2), and QQWSSYPYT (SEQ ID NO: 3), respectively, and
heavy chain complementarity determining regions HCDR 1, IICDR2, and HCDR3
comprising the amino acid sequences GYTFTSRYIH (SEQ ID NO: 4),
WIYPVTGDTYYNEICFKG (SEQ ID NO: 5), and GGGMFYY (SEQ ID NO: 6), for:
(a) detecting and/or quantifying human c-Met in or on a human cell;
(b) detecting and/or quantifying c-Met expressing or overexpressing tumor
cells in a patient;
(c) detecting and/or quantifying c-Met expressing or overexpressing
circulating tumor cells in a blood sample of a patient;
(d) detecting and/or quantifying c-Met expressing or overexpressing tumor
cells in a bodily fluid from a cancer patient such as in urine, ascites fluid,
lymphatic fluid,
spinal fluid, bronchial fluid, blood serum or blood;
(e) assessing whether an individual has, or is at risk for developing, cancer
of a tissue or organ wherein c-Met is expressed or over-expressed;
(f) identifying a patient having a tumor suitable for treatment with an anti-
c-Met therapeutic;

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
7
(g) determining response to treatment with anti-c-Met therapeutic antibody
or chemotherapeutic agent; and/or
0i) treating cancer.
Preferably, with respect to (a)-(d) above, the detecting or quantifying is
carried out
in vitro. More preferably, with respect to (a)-(d) above, the detecting and
quantifying are
are each performed in vitro. Also, with respect to (g) and (h) above, the
treatment
preferably includes the administration of an anti-c-Met therapeutic antibody
such as an
anti-c-Met antibody described in, for example, WO 2010/059654 and/or U.S.
Patent No.
8,217,148, and/or an anti-c-Met chemotherapeutic agent such as the compound,
or
pharmaceutically acceptable salt thereof, disclosed in WO 2010/011538 and U.S.
Patent
Application Publication No. 2012/0028984. More preferably, with respect to (g)
and (h)
above, the treatment includes the administration of an anti-c-Met therapeutic
antibody, or
antigen-binding fragment thereof, that specifically binds to the extracellular
domain
(ECD) of human c-Met consisting of the amino acid sequence as in SEQ ID NO:
19, the
antibody, or fragment thereof, comprising:
light chain complementarity determining regions LCDR1, LCDR2, and LCDR3
comprising the amino acid sequences SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID
NO:
24, respectively, and
heavy chain complementarity determining regions HCDR1, HCDR2, and HCDR3
comprising the amino acid sequences SEQ ID NO: 25, SEQ ID NO: 26, and SEQ ID
NO:
27, respectively.
Even more preferably, with respect to (g) and (h) above, the treatment
includes the
administration of an anti-c-Met therapeutic antibody, or antigen-binding
fragment thereof,
comprising a light chain variable region (LCVR) comprising the amino acid
sequence as
in SEQ ID NO: 28, and a heavy chain variable region (HCVR) comprises the amino
acid
sequence as in SEQ ID NO: 29.
Even more preferably, with respect to (g) and (h) above, the treatment
includes the
administration of an anti-c-Met therapeutic antibody, or antigen-binding
fragment thereof,
comprising a light chain comprising the amino acid sequence as in SEQ ID NO:
30, and a
heavy chain comprising the amino acid sequence as in SEQ ID NO: 31 or 32.
Even more preferably, with respect to (g) and (h) above, the treatment
includes the
administration of an anti-c-Met therapeutic antibody comprising two light
chains and two

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
8
heavy chains, wherein each of the light chains consist of the amino acid
sequence as in
SEQ ID NO: 30 and each of the heavy chains consist of the amino acid sequence
as in
SEQ ID NO: 31 or 32 (an human IgG2 or human IgG4 subtype, respectively, of the
anti-
c-Met therapeutic antibody referred to as C8-H241 in WO 2010/059654 and/or
U.S.
Patent No. 8,217,148) or the chemotherapeutic agents shown below as Structure
1 and
Structure 2, or pharmaceutically acceptable salts thereof.
Structure 1: N-(3- Fluoro-4-( I -methy1-6-(1H-pyrazol-4-y1)- H-indazol-5-
y loxy)ph eny1)-1-(4-11u oroph eny1)-6-methy1-2-oxo-1,2-dihy dropy ri dine-3-
carbo xamide
H3
H3C¨N NN
0 0
0
N¨ N
Structure 2: 6-(1-Methy1-1H-pyrazol-4-y1)-3-(2-methyl-2H-indazol.-5-ylthio)-
[1,2,4]triazoto[4,3-b]pyridazine
H3
Nki
HC3
N
/1\1õN N
N
Another aspect of this invention provides isolated nucleic acid molecules
encoding the anti-e-Met antibodies of the invention, expression vectors
comprising the
nucleic acid molecules, and host cells comprising the nucleic acid molecules,

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
9
Figure 1 shows FACS analysis of mAb OptD11 binding to cell surface c-Met in
different tumor cells.
Figure 2 shows, in graph format, a binding curve for soluble c-Met ECD-Fc
fusion
protein by solid-phase ELISA using the anti-c-Met therapeutic antibody C8-H241
(human
IgG4 subtype) as a c-Met capture antibody and biotinylated rnAb OptD11 (murine
IgG1
subtype) as a c-Met detection antibody reagent.
Figure 3 illustrates the simultaneous but non-competitive binding of m.Ab C8-
H241 (human IgG4 subtype) and OptD11 to human c-Met-ECD-Fc-Flis fusion protein
as
determined by use of a surface plasmon resonance biosensor.
Definitions
As used herein, "c-Met" or "human c-Met" refers to any human c-Met, unless
otherwise indicated, as well as functionally active, mutated forms thereof.
The structure
of c-Met is depicted schematically as:
Extracellular Domain (ECM Intracellular Domain
SEMA PSI -- 4 IPT -- TM JM ICD intracellular tail
SEMA: Sema domain
PSI: Plexin, Semaphorins, and Integrins domain
IPT: 4 Imimmoglobulins, Plexins, and Transcription factor
domains
TM: Transmembrane region
3M: Juxtamembrane domain
KD: Kinase domain
in the human c-Met ECD (SEQ ID NO: 19), amino acids 1-24 comprise the signal
sequence. The mature protein begins at amino acid 25 of SEQ ID NO: 19. The
SEMA
domain consists of approximately 500 amino acid residues at the N-terminus of
c-Met,
and contains the a-chain (amino acid residues 25-307 of SEQ ID NO: 19, i.e.,
(SEQ ID

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
NO: 20) and part of the n-chain (amino acid residues 308-519 of SEQ ID NO: 19,
i.e.,
(SEQ ID NO: 21)).
As used herein, the term "antibody" refers to a monoclonal antibody, unless
otherwise indicated. The term "monoclonal antibody", its abbreviation "mAb",
and
5 grammatical forms thereof are intended to refer to antibodies that are
derived from a
single copy or clone including, for example, any eukaryotic, prokaryotic, or
phage clone,
and not the method by which it is produced. Monoclonal antibodies of the
invention
preferably exist in a homogeneous or substantially homogeneous population.
Complete
inAbs contain two heavy chains and two light chains. "Antigen-binding
fragments" of
10 such monoclonal antibodies include, for example, Fab fragments, Fab'
fragments, F(ab)2
fragments, and single chain Fv fragments. Monoclonal antibodies and antigen-
binding
fragments thereof of the invention can be produced, for example, by
recombinant
technologies, phage display technologies, synthetic technologies, e.g., CDR-
grafting, or
combinations of such technologies, or other technologies known in the art. It
is
understood that regardless of whether antigen-binding fragments thereof are
specified, the
terms "antibody" and "monoclonal antibody" as used herein includes such
fragments as
well as single chain forms, unless indicated otherwise.
Methods for producing and purifying monoclonal antibodies and antigen-binding
fragments thereof are well known in the art and can be found, for example, in
Harlow and
Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory
Press,
Cold Spring Harbor, New York, chapters 5-8 and 15, ISBN 0-87969-314-2.
The c-Met antibodies disclosed herein are useful for detecting expression,
over-
expression, or the level of c-Met present in or on cells, or in or on cells in
tissues, organs,
bodily fluids, etc., and in diagnostic, prognostic, and/or patient monitoring
procedures.
The term "bodily fluid" refers to any fluid or other material derived from the
body of a
normal or diseased subject, such as blood, blood serum, plasma, lymph, bone
marrow,
urine, saliva, tears, cerebrospinal fluid, milk, amniotic fluid, bile, urine,
bronchial fluid,
ascites fluid, pus, or any other biological product. Also included within the
meaning of
this term is an organ or tissue extract, and a culture fluid in which any
cells or tissue
preparation from a subject have been incubated.
The phrase "specifically binds" as used herein in reference to the affinity of
a

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
11
c-Met antibody for the ECU of human c-Met is intended to mean, unless
indicated
otherwise, a KD of less than about 1 x 10-8 M, preferably, less than about 1 x
104 M,
more preferably, between about 1 x las M and about 1 x 1041 M, even more
preferably,
between about 1 x 104 M and about 5 x 10-11 M, even more preferably, between
about 5 x
10-1 M and about 5 x 10-11 M, even more preferably, between about 0.15 nM and
about
0.05 nM, even more preferably, between about 0.15 nM and about 0.075 nM, and
most
preferably, about 0.10 nM, as determined by common methods known in the art,
including by use of a SPR biosensor essentially as described herein.
The term "tumor" refers to all neoplastic cell growth and proliferation,
whether
malignant or benign, and all pre-cancerous and cancerous cells and tissues.
The terms "cancer" and "cancerous" refer to or describe the physiological
condition in mammals typically characterized by aberrant cell
growth/proliferation.
Examples include but are not limited to: carcinoma, lymphoma, blastoma,
sarcoma, and
leukemia. The terms "cancer", "cancerous", and "tumor" are not mutually
exclusive as
used here.
As used herein, human c-Met is "over-expressed" in or on a human cell, CTC, or
tumor tissue sample when the quantity of human c-Met is determined to be
significantly
greater for the human cell, CTC, or tumor tissue sample than the quantity of
human c-Met
in normal human cells or non-tumor human tissue.
Antibody Compositions and Methods
The c-Met monoclonal antibodies of the invention target c-Met in neoplasms
regardless of tissue of origin. Because of the relatively greater expression
of c-Met on
tumor cells, it is possible to distinguish tumors from normal tissue. Also,
because of the
broad expression of c-Met across tumor classes (i.e., different organs and
tissues of
origin), imaging of c-Met as a surface marker is not specific to any
particular tumor type,
but can be generally used for any c-Met-expressing tumor.
There are well-known methods in the art that a skilled artisan may use to form
stable, detectable antigen-antibody complexes (see, e.g., Antibodies, A
Laboratory
Manual by Harlow and Lane (current edition), Cold Spring Harbor Laboratory
Press,
Cold Spring Harbor, New York, for conditions permitting formation of
detectable

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
12
antigen/antibody. complexes). In particular, WO 2010/059654 and/or U.S. Patent
No.
8,217,148 describes exemplary conditions which may permit the anti-c-Met
monoclonal
antibodies, or antigen-binding fragments thereof, of the present invention
including, but
not limited to the monoclonal antibody referred to herein as OptD11. A
composition
comprising an anti-c-Met monoclonal antibody of the present invention bound to
the ECD
of c-Met may also be detected, labeled, and/or identified using methods taught
herein or
generally known in the art, including, but not limited to, such methods
disclosed in
Harlow and Lane, ibid, or WO 2010/059654.
The anti-c-Met monoclonal antibodies of the present invention or the c-
Met/anti-
c-Met monoclonal antibody complexes described herein can be detectably labeled
using
any art-known means (see, e.g., Antibody Engineering Volume 2, Kontermann,
Roland;
Dilbel, Stefan (Eds.)). Labels can be, for example, without limitation, light-
emiting or
light-absorbing agents, chromophores, chromogens, magnetic or iron particles,
dyes,
fluorescents, fluorophores, phosphorescents, chemiluminescents,
bioluminescents agent,
radionuclides, enzymes, positron emission tomographic-imageable agents,
magnetic
micro-beads, ferrofluid nanoparticles, secondary antibodies, and magnetic
resonance-
imageable agents.
Without limitation, c-Met or a detectably labeled c-Met/anti-c-Met monoclonal
antibody complex can be on a cell, or fragment thereof, either in vivo, ex
vivo or in vitro.
For example, without limitation, such a cell or fragment thereof can be, in
situ, isolated
from its naturally occurring state, or in a sample, such as, e.g., from a cell
pellet,
xenograft, tissue (cancerous or non-cancerous), organ, bodily fluid, or any
concentrated,
purified, enriched form thereof. In any of these methods, the contacting and
detecting can
each be performed in vitro, the contacting can be performed in vivo and the
detecting can
be performed in vitro (or vice versa), or the contacting and the detecting can
each be
performed in vivo.
The term "detectably labeled" means that the anti-c-Met antibody, or antigen-
binding fragment thereof of the present invention, or a complex of c-Met/anti-
c-Met
monoclonal antibody has attached to it, either covalently or non-covalently, a
useful
detectable label. In direct conjugate-labeled antibody methods, many different
useful
labels can be employed including, for example, prosthetic group complexes,
chromophores, chromogens (color-producing substrates), dyes, fluorescent
compounds,

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
13
fluorogenic compounds, radioactive isotopes, paramagnetic isotopes, and
compounds that
can be imaged by positron emission tomography (PET) and magnetic resonance
imaging
(MR1). Other suitable labels are art-known or can be determined by routine
experimentation. In indirect methods, a secondary antibody can be conjugated
with, for
example, an enzyme. Binding of the secondary antibody to the primary antibody,
which
is bound to the target antigen, can then be detected by reaction with a
chromogenic
substrate of the enzyme under appropriate conditions to yield a detectable
signal.
Colorimetric detection can be used, employing chromogenic compounds that
have, or result in, chromophores with high extinction coefficients, and which
are
therefore easily detectable. When later exposed to its substrate under
appropriate reaction
conditions, the enzyme will react with the substrate to produce a chemical
moiety that can
be detected, for example, by spectrophotometric, fluorometric, or visual
means.
Enzymes commonly used for this purpose include horseradish peroxidase,
alkaline
phosphatase, glucose-6-phosphate dehydrogenase, malate dehydrogenase,
staphylococcal
nuclease, s-V-steroid isomerase, yeast alcohol dehydrogenase, a-
glycerophosphate
dehydrogenase, triose phosphate isomerase, asparaginase, glucose oxidase,p-
galacto-
sidase, ribonuclease, urease, catalase, glucoamylase, and
acetylcholinestentse.
Examples of suitable prosthetic group complexes include, e.g., without limit,
streptavidin/biotin and avidinibiotin.
Commonly used chromogens include diaminobenzidine (DAB); DA.B with
enhancement; 3-amino-9-ethyl carbazole (AEC); 4-chloro-l-naphthol (4-CN);
Hanker-
Yates reagent; alpha-naphthol pyronin; 3,3',5,5'-tetramethylbenzidine (TMB);
Fast Blue
BB; Fast Red TR; new fuchsin; BCIP-NBT; tetrazolium; tetranitoblue tetrazolium
(TNBT); and immunogold with silver enhancement.
Use of chromogens is preferred because assays employing them can be easily
performed in clinical diagnostic laboratories and reviewed by a pathologist
with
equipment commonly available in these laboratories.
An anti-c-Met monoclonal antibody herein conjugated to an enzyme can also be
used in an enzyme-linked immunosorbent assays (ELISAs). Such assays are
described in
detail in, for example, Butler (1994) "ELISA" (Chapter 29), In: van Oss, C. J.
et al., eds.,
Immunochemistr y, Marcel Dekker, Inc., New York, pp. 759-803. The present c-
Met

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
14
antibodies can also be used in radioimmunoassay and fluorescence-activated
cell sorting
(FACS) analysis of cell-surface c-Met.
Useful fluorescent labels include umbelliferone, fluorescein, fluorescein
isothiocyanate, dichlorotriazinylamine fluorescein, rhodamine, a dansyl group,
phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde, fluorescamine,
and Cy5
(Haugland ((1996) Handbook of Fluorescent Probes and Research Chemicals, Sixth
Ed.,
Molecular Probes, Eugene, OR).
The anti-c-Met antibodies, or antigen-binding fragments thereof, or c-Met/anti-
c-
Met monoclonal antibody complexes of the present invention can also be
detectably
labeled using fluorescence-emitting metals such as 152Eu+, or other members of
the
lanthanide series, by attaching them using such metal chelating groups as
diethylenetriaminepentaacetic acid (DTPA) or ethylenediamine-tetraacetic acid
(EDTA).
The anti-c-Met antibodies, or antigen-binding fragments thereof, or c-Metlanti-
c-
Met monoclonal antibody complexes of the present invention can also be
detectably
labeled by coupling them to a phosphorescent or chemiltuninescent compound
that can
then be detected by the phosphorescence or luminescence that arises during the
course of
a chemical reaction. Examples of useful chemiluminescent compounds include
luminol,
isoluminol, theromatic acridinium ester, imidazole, acridinium salt, and
oxalate ester.
Likewise, a bioluminescent compound such as luciferin, luciferase, or aequorin
can be
used to label the antibody peptides. The presence of a bioluminescent protein
is
determined by detecting the presence of luminescence.
In vivo or in vitro imaging can be used to detect the proliferation,
migration, or
invasion of c-Met expressing or over-expressing tumor cells (and/or
circulating tumor
cells) in a patient, including occult metastases that are not observable by
other methods.
The expression of c-Met can be correlated with disease progression in cancer
patients;
patients with late stage cancer will usually have higher levels of c-Met
expression in both
their primary tumors and metastases. c-Met-targeted imaging could be used to
stage
tumors non-invasively, or to detect another disease associated with the
presence of
increased levels of c-Met.
The invention encompasses antibodies recombinantly fused or chemically
conjugated (including both covalent and non-covalent conjugations) to a
polypf...ptide (or

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
portion thereof, preferably comprising at least: 10, 20, 30, 40, 50, 60, 70,
80, 90, or 100
contiguous amino acids of the polypeptide) to generate fusion proteins. The
fusion does
not necessarily need to be direct, but may occur through linker sequences.
Antibodies fused or conjugated to a polypeptide may also be used in in vitro
5 immunoassays and in purification methods using known art methods (see,
e.g., Harbor, et
al., supra, and WO 93/2 1232; EP 439,095; Naramura et al. (1994) Imtnunol.
Lett. 39:9 1-
9; U.S. Patent No. 5,474,981; Gillies, et al. 1992 PNAS 89:1428-32; Fell, et
al. 1991 J.
Immunol. 146:2446-52).
The invention further includes compositions comprising a polypeptide (or
10 fragment thereof) fused or conjugated to an antibody domain other than a
variable region.
Methods for fusing or conjugating a polypeptide (or fragment thereof) to an
antibody or
antibody portion are known (see, e.g., U.S. Patent Nos. 5,336,603; 5,622,929;
5,359,046;
5,349,053; 5,447,851; 5,112,946; EP 307,434; EP 367,166; WO 96/04388; WO
91/06570; Ashkenazi, etal. (1991) PNAS 88: 10535-10539; Zheng, etal. (1995) J.
15 Immunol. 154:5590-5600; and Vie, et al. (1992) PNAS 89: 11337- 11341).
A polypeptide, polypeptide fragment, may be fused or conjugated to an antibody
or antigen-binding fragment thereof described herein to increase the in vivo
half-life.
Further, a polypeptide, polypeptide fragment, may be fused or conjugated to an
antibody
or antigen-binding fragment thereof to facilitate purification. In preferred
embodiments,
the polypeptide is a hexa-histidine peptide, such as the tag provided in a pQE
vector
(QIAGEN, Inc., Chatsworth, CA), among others, many of which are commercially
available. Hexa-histidine provides for convenient purification of a protein
(Gentz, et al.
(1989) PNAS 86:821-824). Other peptide tags useful for purification include,
e.g., the
"HA" tag, which corresponds to an epitope derived from the influenza
hemagglutinin
protein (see, e.g., Wilson, et al. (1984) Cell 37:767-778) and the "flag" tag.
The invention further encompasses antibodies or fragments thereof conjugated
to
a diagnostic or therapeutic agent. The antibodies can be used diagnostically
to, for-
example, monitor the development or progression of a tumor as part of a
clinical testing
procedure to determine the efficacy of a given treatment regimen. Detection
can be
facilitated by coupling the antibody to a detectable label. The detectable
label may be
coupled or conjugated either directly to the antibody (or fragment thereof) or
indirectly,
through an intermediate (such as, e.g., an art known linker) using established
techniques

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
16
(see, e.g., U.S. Patent No. 4,741,900 for metal ions that can be conjugated to
antibodies
for use as diagnostics according to the invention).
A monoclonal antibody, or antigen-binding fragment thereof, of the invention
can
also be attached to solid supports, which are particularly useful for
immunoassays or
purification of a target antigen. Such solid supports include, e.g., without
limitation,
glass, cellulose, poly-acrylamide, nylon, polystyrene, polyvinyl chloride, or
polypropylene. Techniques for conjugating a therapeutic moiety to an antibody
are
known, see, e.g., Amon, et al., "Monoclonal Antibodies For immunotargeting Of
Drugs
In Cancer Therapy", in Monoclonal Antibodies And Cancer Therapy, Reisfeld, et
al.
(eds.), pp. 243-56 (Alan R. Liss, Inc.1985); Tiellstrom, et al., "Antibodies
For Drug
Delivery", in Controlled Drug Delivery (2nd Ed.), Robinson, et al. (eds.), pp.
623-53
(Marcel Dekker, Inc. 1987); Thorpe, "Antibody Carriers Of Cytotoxic Agents In
Cancer
Therapy: A Review", in Monoclonal Antibodies '84: Biological And Clinical
Applications, Pinchera, et al. (eds.), pp. 475-506 (1985); "Analysis, Results,
and Future
Prospective of the Therapeutic Use Of Radiolabeled Antibody in Cancer
Therapy", in
Monoclonal Antibodies for Cancer Detection and Therapy, Baldwin, et al.
(eds.), pp. 303-
16 (Academic Press 1985), and Thorpe, et al., "The Preparation and Cytotoxic
Properties
of Antibody-Toxin Conjugates," Immune'. Rev. 62: 119-58 (1982).
A particular protein such as c-Met can be measured by a variety of immunoassay
methods including, e.g., without limitation, competitive and non-competitive
assay
systems using techniques such as, e.g., without limitation, western blots,
radioirnmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich"
immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion
precipitin
reactions, immunodiffusion assays, agglutination assays, complement-fixation
assays,
immunoradiometric assays, fluorescent immunoassays, and protein .A
immunoassays. For
a review of immunological and immunoassay procedures in general, see Stites
and Ten
(eds.) (1991) Basic and Clinical Immunology (7th ed.). Moreover, the
immunoassays of
the invention can be performed in many configurations, which are reviewed
extensively
in Maggio (ed.) (1980) Enzyme Immunoassay CRC Press, Boca Raton, Florida;
Gosling .1
P 2000 Immunoassays: A Practical Approach (Practical Approach Series) Oxford
Univ
Press; Diamandis & Christopoulus, 1996 Immunoassay Academic Press, San Diego,
CA;
Tijan (1985) "Practice and Theory of Enzyme Immunoassays." Laboratory
Techniques in

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
17
Biochemistry and Molecular Biology, Elsevier Science Publishers B.V.,
Amsterdam;
Wild, D. (Ed.), 2001 The Immunoassay Handbook (2nd edition) Nature Pub Group;
James T. Wu, 2000 Ouantitative Immunoassay: A Practical Guide for Assay
Establishment. Troubleshooting. and Clinical Application, Amer Assn for
Clinical
Chemistry, Brousseau & Beaudet (Eds.) Manual of Immunological Methods CRC
Press
Boca Raton, Florida; and Harlow and Lane Antibodies, A Laboratory Manual,
supra. See
also Chan (ed.) (1987) Immunoassay: A Practical Guide Academic Press, Orlando,
FL;
Price and Newman (eds.) (1991) Principles and Practice of Immunoassays
Stockton
Press, NY; and Ngo (ed.) (1988) Non-isotopic Immunoassays Plenum Press, NY.
Immunoassays for measurement can be performed by a variety of art-known
methods. In brief, immunoassays to measure the c-Met LCD can be either
competitive or
noncompetitive binding assays. In competitive binding assays, the sample to be
analyzed
competes with a labeled analyte for specific binding sites on a capture agent
bound to a
solid surface. Preferably, the capture agent is an antibody specifically
reactive with the c-
Met ECD as described herein. The concentration of labeled analyte bound to the
capture
agent is inversely proportional to the amount of five analyte present in the
sample.
In a competitive binding immunoassay, the target protein present in the sample
(i.e., the c-Met LCD) competes with labeled protein for binding to an
antibody, or
antigen-binding fragment thereof, of the present invention. The antibody, or
antigen-
binding fragment thereof, of the present invention may be bound to a solid
surface to
effect separation of bound-labeled protein from the unbound-labeled protein.
Alternately,
the competitive binding assay may be conducted in liquid phase and a variety
of
techniques known in the art may be used to separate the bound-labeled protein
from the
unbound-labeled protein. Following separation, the amount of bound labeled
protein is
determined. The amount of protein present in the sample is inversely
proportional to the
amount of labeled protein binding.
Alternatively, a homogeneous immunoassay may be performed in which a
separation step is not needed. In these immunoassays, the label on the protein
is altered
by the binding of the protein to its specific binding composition. This
alteration in the
labeled protein results in a decrease or increase in the signal emitted by
label, so that
measurement of the label at the end of the immunoassay allows for detection or
quantitation of the protein.

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
18
Competitive assays are also particularly useful, where the cells are contacted
and
incubated with a labeled antibody having known binding affinity to the
protein, such as
'251-antibody, and a test sample whose binding affinity to the binding
composition is
being measured. The bound and free-labeled binding compositions are then
separated to
assess the degree of protein binding. The amount of test compound bound is
inversely
proportional to the amount of labeled binding partner binding to the known
source. Any
one of numerous techniques can be used to separate bound from free protein to
assess the
degree of protein binding. This separation step could typically involve a
procedure such
as adhesion to filters followed by washing, adhesion to plastic followed by
washing, or
centrifugation of the cell membranes. Viable cells could also be used to
screen for the
effects of drugs on a c-Met mediated function (e.g., second messenger levels,
such as,
e.g., cell proliferation; inositol phosphate pool changes, transcription using
a luciferase-
type assay; and others). Some detection methods allow for elimination of a
separation
step, e.g., a proximity-sensitive detection system.
Qualitative or quantitative analysis of c-Met may also be determined by a
variety
of noncompetitive immunoassay methods using the antibodies, or antigen-binding
fragments thereof, of the present invention. For example, a two-site, solid
phase
sandwich immunoassay may be used. In this type of assay, an antibody is
attached to a
solid support. A second protein-binding composition, which may also be an
antibody,
and which binds the protein at a different site, is labeled. After binding at
both sites on
the protein has occurred, the unbound-labeled binding composition is removed
and the
amount of labeled binding composition bound to the solid phase is measured.
The
amount of labeled binding composition bound is directly proportional to the
amount of
protein in the sample.
The ability of the antibody of interest to immtmoprecipitate a particular
antigen
can be assessed by, e.g., Western blot analysis. One of skill in the art would
be
knowledgeable as to the parameters are modifiable to increase binding of an
antibody to
an antigen and to decrease background (e.g., by pre-clearing the cell lysate
with sepharose
beads). Further discussion of immunoprecipitation protocols can be found in,
e.g.,
Ausubel et al, eds., 1994, Current Protocols in Molecular Biology, Vol. 1,
John Wiley &
Sons, Inc., New York.

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
19
An EL1SA assay comprises preparing an antigen, coating the well of a 96 well
microtiter-plate with the antigen, adding the antibody of interest conjugated
to a
detectable compound such as an enzymatic substrate (e.g., horseradish
peroxidase or
alkaline phosphatase) to the well and incubating for a period of time, and
detecting the
presence of the antigen. In ELISAs, the antibody of interest does not have to
be
conjugated to a detectable compound; instead, a second antibody (which
recognizes the
antibody of interest) conjugated to a detectable compound may be added to the
well.
Further, instead of coating the well with the antigen, the antibody may be
coated to the
well. In this case, a second antibody conjugated to a detectable compound may
be added
following the addition of the antigen of interest to the coated well. An
ordinary artisan
can determine without undue experimentation what parameters to adjust, e.g.,
to increase
signal as well as what other variations for an ELISA should be used (see,
e.g., Ausubel, et
al., eds., 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley &
Sons, Inc.,
New York). For example, OptD11 may be useful as a c-Met ECD detection antibody
for
measuring total c-Met ECD in human tumors, tumor lysates, and bodily fluids
such as
blood if used in conjunction with a c-Met ECD capture antibody such a c-Met
monoclonal antibody known in the art including, but not limited to, the c-Met
monoclonal
antibody known in the art as C8-H241 (see, for example, Chemical Abstracts
Service
#1365287-97-3). Alternatively, OptD11 may be useful as a c-Met ECD capture
antibody
for measuring total c-Met ECD in human tumors, tumor lysates, or bloodily
fluids such as
blood if used in conjunction with a c-Met ECD detection antibody such a c-Met
monoclonal antibody known in the art including, but not limited to, the c-Met
monoclonal
antibody known in the art as C8-H241. Furthermore, OptD11 may be useful as a c-
Met
ECD capture antibody fur measuring phosphor-c-Met in human tumors, tumor
lysates, or
bodily fluids such as blood if used in conjunction with a detection antibody
such as an
anti-phospho-tyrosine antibody conjugated to HRP (e.g., R&D Systems,
Minneapolis,
MN; catalog #841403) or an anti-phospho-c-Met pYpYpY1230/1234/1235 c-Met
polyclonal antibody (e.g., Invitrogen, Carlsbad, CA; catalog #44-8880).
The binding affinity of an antibody to an antigen and the on- and off-rate of
an
antibody-antigen interaction can be determined by, e.g., using a competitive
binding
assay. One non-limiting example is a radioimmunoassay comprising incubating
labeled
antigen (e.g., using 3H or 1250 with an antibody of interest in the presence
of increasing

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
amounts of unlabeled antigen, and then detecting the amount of antibody bound
to the
labeled antigen. The affinity of the antibody of interest for a particular
antigen and the
binding off-rates can be determined from the data by, e.g., Scatchard plot
analysis.
Competition with a second antibody can also be determined using, e.g.,
5 radioimmunoassays.
Useful radiolabels, which are detected simply by gamma counter, scintillation
1241 125, 131 35 14
counter, PET scanning, or autoradiography, i 3
nclude H, 1, S , and
, , C. For
in vivo diagnosis, radionuclides can be bound to the rnAb or antigen-binding
fragments
either directly or indirectly using a chelating agent such as DIPA and EDTA.
Examples
99 123 125 131 111 97 67 67 68 72
10 of such radionuclides include Tc, 1, 1, I, In, Ru, Cu, Ga, Ga, As,
89 90
9 Y and 201T1. Generally, the amount of labeled mAb needed for detectability
in in
vivo diagnostic use will vary depending on the patient's age, condition, sex,
extent of
disease, contraindications, if any, and other variables, and can be readily
adjusted by the
attending physician or diagnostician. A preferred diagnostic method is
radioimmuno-
15 scintigraphic analysis, which is preferably performed in a manner that
results in serial
total body gamma camera images, and allows determination of regional activity
by
quantitative "region-of-interest" (ROI) analysis.
in another embodiment, the monoclonal antibodies of the present invention may
be used to detect circulating tumor cells (CTCs). CTCs are tumor cells that
are shed from
20 tumors, survive within the bloodstream during transit and initiate a new
growth in distant
sites. Detecting CTCs is useful as CTCs can be found in patients before a
tumor is
detected. Circulating tumor cells are also found in a significant proportion
of patients
when a carcinoma recurs, and CTCs persist in some patients after removal of
the primary
tumor. Evidence suggests that CTCs are derived from clones in the primary
tumor as
well as metastatic tumors and that they may reflect the tumor burden at all
stages of tumor
progression. Thus, in addition to a potential role in early diagnosis and
prognostication,
CTCs may play a major role in characterizing genetic and phenotypic changes
with tumor
progression, thereby helping to guide targeted therapy. More particularly, the
anti-c-Met
monoclonal antibodies of the present invention may be useful in assays that
can capture,
identify, and/or quantify CTCs such as, e.g., the CellSearchg CTC Test
(Veridex LLC,
San Diego, CA), Magnetic Activated Cell Sorting System (MACS, Miltenyi Biotec

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
21
GmbH, Germany), Dynal Magnetic Beads* (Invitrogen), EasySep* (Stern Cell
Technologies, Vancouver Canada), CTC Chips (On-Q-ity, Waltham, MA), or any
other
test known in the art for the isolation and detection of CTCs such as those
described in
Sleijfer, et al. Circulating tumour cell detection on its way to routine
diagnostic
implementation? Fur .1 Cancer, 43 (18):2645-50 (2007); Lacroix M.,
Significance,
detection and markers of disseminated breast cancer cells. Endocr Relat
Cancer., 13
(4):1033-67 (2006); and Pante', et al., Detection, clinical relevance and
specific
biological properties of disseminating tumour cells. Nat Rev Cancer, 8 (5):329
¨40
(2008)). Presently, CellSearch* CTC is the only diagnostic test approved by
the USFDA
as an automated test to detect and enumerate circulating tumor cells (Fed.
Reg.
69(91):26036-38 (2004). Results from CellSearch tests have been used to
monitor
disease progression and therapeutic efficacy in metastatic prostate (Danila,
et al.,
Circulating tumor cell number and prognosis in progressive castration-
resistant prostate
cancer. Clin Cancer Res., 13(23):7053-58 (2007)), colorectal (Cohen, et al.,
Isolation
and characterization of circulating tumor cells in patients with metastatic
colorectal
cancer. Clin Colorectal Cancer 6 (2):125-32 (2006)), and breast
(Cristofanilli, et al.
Circulating tumor cells, disease progression, and survival in metastatic
breast cancer. N
Engl i Med., 351(8):781-91 (2004)) cancer. The anti-c-Met antibody, or antigen-
binding
fragment thereof, of the present invention, e.g., OptD11, can be used in such
methods,
e.g.. CellSearch* tests, and in some cases performed at the start of therapy
and any time
during the course of treatment for a c-Met-mediated-cancer. Preferably, an
anti-c-Met
antibody, or antigen-binding fragment thereof, of the present invention may be
used in
such methods along with antibodies specific for other polypeptides including,
but not
limited to, EPCAM, DAPI, CD45, and/or cytokeratin (including, but not limited
to
cytokeratin 7, 8, 18, and/or 19). Information generated from such testing may
be useful
for its prognostic value by allowing, e.g., monitoring of disease progression
and
therapeutic efficacy and may allow earlier (and ongoing) treatment decisions.
Further, by
permitting simultaneous binding of a detectably labeled antibody of the
present invention
and a therapeutic anti-c-Met antibody (such as disclosed in WO 2010/059654
and/or U.S.
Patent No. 8,217,148) a break in treatment with a therapeutic anti-c-Met
antibody, for
example, "washing out" the therapeutic antibody to allow the diagnostic
antibody to bind
c-Met, is not required. Consequently, an anti-c-Met antibody of the invention
permits

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
22
uninterrupted therapeutic treatment concomitantly with diagnostic monitoring,
as
necessary.
For in vivo applications, the detectably-labeled, anti-c-Met antibodies, or
antigen-
binding fragments thereof, of the invention can be formulated in convenient
forms for
administration. For diagnosis, the detectably labeled anti-c-Met antibodies,
or antigen-
binding fragments thereof, of the invention can be administered systemically,
e.g.,
parenterally, by injection or infusion. Such injection or infusion can be by
any known
route, preferably intravenous injection or infusion, subcutaneous injection,
intramuscular,
intracranial, or intrathecal injection or infusion, or intraperitoneal
administration.
Injectables can be prepared in conventional forms, either as solutions or
suspensions, or
as solid forms. Such compositions can be prepared by methods well kn.own in
the art.
See, e.g., Remington: The Science and Practice of Pharmacy, 19th ed. (1995),
A. Gennaro
et al., Mack Publishing Co., and comprise anti-c-Met antibodies disclosed
herein, and a
pharmaceutically or diagnostically acceptable carrier, diluent, or excipient.
Dosage can vary from 0.01 mg/kg to 100 mg/kg body weight. Further guidance
regarding appropriate doses for diagnostic imaging can be found in Smith et
al. (1977)
Antibodies in Human Diagnosis and Therapy, Haber et al., eds., Raven Press,
New York,
pages 365-389.
For in vivo c-Met detection purposes, the term "effective amount" of the
present
anti-c-Met monoclonal antibody, or antigen-binding fragment thereof, refers to
the
amount of antibody compound which, upon single or multiple dose administration
to a
patient, provides the ability to detect c-Met in vivo.
Articles of Manufacture and Kits
The invention also provides articles of manufacture and kits containing
compositions useful for diagnosing, detecting, quantifying, and/or imaging c-
Met-positive
tumors and non-cancerous cells. The article of manufacture may comprise a
container
with a label. The container may holds a composition comprising the anti-c-Met
monoclonal antibody, or antigen-binding fragment thereof, of the invention,
which is
either detectably labeled, or unlabeled. The label on the container may
indicate that the
composition is used fur prognosing or monitoring cancer, or for diagnosing or
monitoring
particular types of cancer or tumors that express c-Met or for which c-Met
levels or

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
23
turnover is prognostic, or for prediction of an effective target for therapy.
In another
embodiment, the label may indicate that the composition is useful for
detecting and/or
quantifying c-Met, and can also indicate directions for either in vivo or in
vitro use.
The kit of the invention can also comprise a container comprising a secondary
antibody or even a c-Met antigen, preferably antigen comprises the
extracellular domain
(ECD) of human c-Met consisting of the amino acid sequence as in SEQ ID NO:
19. The
secondary antibody can be conjugated with an enzyme. A chromogenic substrate
of the
enzyme can also be included in the kit. The kit may further include other
materials
desirable from a commercial and user standpoint, including buffers, diluents,
filters,
needles, syringes, and package inserts with instructions for use in vivo, in
vitro, or both.
Aberrant HGF and/or c-Met signaling is inversely correlated with clinical
outcome, and has been documented in a wide range of human malignancies.
Inappropriate
expression of c-Met correlates with poor patient prognosis in many human
tumors. The
present monoclonal antibody and antigen-binding fragments thereof that bind c-
Met
useful in diagnostic procedures can be used to identify and stratify cancer
patients, and to
monitor patient responses to c-Met targeted therapy by using tissue samples
collected by
routine standards, and which impose minimum patient discomfort.
The present methods thus offer newly diagnosed cancer patients a form of
metastatic risk stratification that uses noninvasive means to assess as high
or low the
probability that a given tumor will subsequently invade or metastasize,
without any
dependence on the tumor's tissue of origin. Such information improves the
ability to
design appropriate monitoring and therapy protocols on an individual patient
basis.
The c-Met antibodies or c-Met/anti-c-Met monoclonal antibody complexes of the
present invention and methods disclosed herein can be used in the diagnosis of
a variety
of mammalian species, and are equally applicable in the practice of human or
veterinary
medicine. Thus, these antibodies, complexes, and methods may be used with
domestic
and commercial animals, and most preferably, with humans.
Another aspect of this invention pertains to isolated nucleic acid molecules
encoding any of the aforementioned anti-c-Met antibodies, expression vectors
comprising
the nucleic acid molecules, and host cells comprising the nucleic acid
molecules. The
invention further provides methods of purifying any of the aforementioned anti-
c-Met
antibodies.

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
24
Preferred host cells for transformation of vectors and expression of the
antibodies
of the present invention are mammalian cells, e.g., NSO cells (non-secreting
(0) mouse
myelorna cells), 293, SP20 and CHO cells and other cell lines of lymphoid
origin such as
lymphoma, myeloma, or hybridoma cells. Other eukaryotic hosts, such as yeasts,
can be
alternatively used.
An "isolated" antibody in reference to an anti-c-Met antibody is one that has
been
identified and separated and/or recovered from a component of its natural
environment.
Contaminant components of its natural environment are materials that would
interfere
with diagnostic or therapeutic uses for the antibody, and may include enzymes,
hormones,
and other proteinaceous or nonproteinaceous solutes. In preferred embodiments,
an
antibody of the present invention will be purified (1) to greater than 95% by
weight of
antibody as determined by the Lowry method, and most preferably more than 99%
by
weight, (2) to a degree sufficient to obtain at least 15 residues of N-
terminal or internal
amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity
by SDS-
PAGE under reducing or nonreducing conditions using Coomassie blue or,
preferably,
silver stain. The term "isolated" in reference to an anti-c-Met antibody of
the present
invention may include the antibody in situ within recombinant cells since at
least one
component of the antibody's natural environment will not be present. The anti-
c-Met
antibodies of the present invention may be isolated or purified by any method
known in
the art, including precipitation by ammonium sulfate or sodium sulfate
followed by
dialysis against saline, ion exchange chromatography, affinity or immuno-
affinity
chromatography including, but not limited to, Protein-A affinity
chromatography, as well
as gel filtration or zone electrophoresis.
Additionally, the present invention provides expression vectors containing the
polynucleotide sequences previously described operably linked to a control
sequence such
as an expression sequence, a promoter and/or an enhancer sequence. A variety
of
expression vectors for the efficient synthesis of antibody polypeptide in
prokaryotic
systems, such as bacteria and eukaryotic systems, including but not limited
to, yeast and
mammalian cell culture systems have been developed. The vectors of the present
invention can comprise segments of chromosomal, non-chromosomal and synthetic
DNA
sequences.

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
The present invention also provides recombinant host cells containing the
recombinant vectors previously described. Antibodies of the present invention
can be
expressed in cell lines other than in hybridomas. Nucleic acids, which
comprise a
sequence encoding an antibody according to the invention, can be used for
transformation
5 of a suitable mammalian host cell.
Cell lines of particular preference are selected based on high levels of
expression,
constitutive expression of protein of interest and minimal contamination from
host
proteins. Mammalian cell lines available as hosts for expression are well
known in the art
and include many immortalized cell lines, such as but not limited to, COS-7
cells,
10 Chinese Hamster Ovary (CI-TO) cells, Baby Hamster Kidney (BHK) cells and
many
others including cell lines of lymphoid origin such as lymphoma, myeloma, or
hybridoma
cells.

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
26
Example I
Antibody Expression and Purification
The anti-c-Met antibodies of the present invention, including, but not limited
to
OptD11, may be transiently expressed in HEK293 EBNA cells (Edge BioSystems,
Gaithersburg, MD) using standard transfection procedures. Transfected cells
are cultured
in standard serum-free medium containing geneticin (G418) and tobramycin for
48 to 120
hours at 37 C after transfection. The anti-c-Met antibody may be purified on
a 60 ml
rProtein A Sepharose column (for example, GE Healthcare, Piscataway, NJ;
catalog #17-
1279-04) by following the manufacturer's instructions, and further
concentrated and
purified by size exclusion chromatography (XK50/60 Superdex200, GE Healthcare)
with
phosphate buffered saline (PBS), pH 7.4, as the mobile phase. Next, the
antibody
preparation may be filtered using a Millev-GV, PVDF membrane, 0.22 pm, 33 mm,
(Millipore; #SLGV033RS) and stored at 4 to 8 C.
Table 1. CDR Amino Acid Sequences of Antibody OptD11
CDRs Amino Acid Sequences
OptD11 LCDR. 1 SVSSSISSINLH (SEQ ID NO: 1)
OptD11 LCDR 2 GTSNLAS (SEQ ID NO: 2)
OptD11 LCDR 3 QQWSSYPYT (SEQ ID NO: 3)
OptD11 HCDR 1 GYTFTSRYIH (SEQ ID NO: 4)
OptD11 HCDR 2 WIYPVTGDTYYNEKFKG (SEQ ID NO: 5)
OptD1.1. HCDR 3 GGGMFYY (SEQ ID NO: 6)
Example 2
Binding. Kinetics and iikflinity of .Antibody Opt131 I
The extracellular domains (ECDs) of human and cynomolgus monkey c-Met
sequences may be expressed as Fe fusion proteins with a flag- and His-tag
(Flis-tag) at
the C-terminus of the Fe, as in SEQ ID NOs:17 and 18. These c-Met LCD Fe
fusion

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
27
proteins may be transiently expressed in HEK293 EBNA separately and purified
essentially as described in Example 1.
The binding kinetics of an anti-c-Met monoclonal antibody of the invention to
human and/or cynomolgus monkey c-Met ECDs may be determined by use of a
surface
plasmon resonance biosensor such as a BlAcore 2000, BlAcore 3000, or a
BlAcore
T100 (GE Health Care, Piscataway, NJ) according to methods known in the art.
Except
as noted, all reagents and materials may be purchased from Biacore , and
measurements
may be performed at 25 'C.
Briefly described, the anti-c-Met monoclonal antibody of the invention may be
dissolved in HBS-EP buffer (150 mM sodium chloride, 3 mM EDTA, 0.005% (w/v)
surfactant P-20, and 10 mM N-2-hydroxyethylpiperazine-N'-2-ethanesulfonic acid
(HEPES) at pH 7.4; #BR-1001-88). Goat anti-mouse Fe antibody is immobilized on
flow
cells 1 to 4 of a CM5 sensor chip at a level of 4000 response units (RUs)
using amine
coupling chemistry to capture anti-c-Met antibodies.
Binding may be evaluated using multiple analytical cycles. Each cycle is
performed at a flow rate of 50 pl/minute, and may consist of the following
steps: injection
of about 10 ill of an anti-c-Met monoclonal antibody at 10 Lig/m1 aiming at a
capture of
40-100 RUs, injection of 250 pi of human or cynomolgus monkey c-Met-Flis-Fc
ECD
(starting at 100 nM and using two-fold serial dilutions for each cycle)
followed by 20
minutes for dissociation, and regeneration using about 30 pi of 10 mM glycine
hydrochloride, pH 1.5. Association and dissociation rates for each cycle may
be
evaluated using a "1:1 (Langmuir) binding" model in the BIAevaluation software
4.1.
The anti-c-Met monoclonal antibody of the present invention, OptUll, was
tested
for its binding kinetics to the ECD of human and cynomolgus monkey c-Met
expressed as
Fe fusion proteins with a flag- and His-tag (Flis-tag) at the C-terminus of
the Fe, as in
SEQ ID NOs:17 and 18. As shown in Table 2, OptD11 binds human and cynomolgus
monkey c-Met-Flis-Fc ECUs with very high binding affinity (Kd). Table 2 also
displays
Icon and koff rate data.

CA 02843771 2014-01-30
WO 2013/043452 PCT/US2012/055057
28
Table 2. Binding Kinetics and Affinity of Antibody OptD11
ECD kon (105 1./Ms) kof (1(I4 ifs) Kti (nM)
Human c-Met 1.6 0.5 0.09 0.12 0.12 0.07
Cynomolgus
1.3 0.3 0.47 0.14 0.35 0.04
Monkey c-Met
Example 3
Detection of c-Met on Cell Surfaces by FACS Analysis Using mAb OptD11
An in vitro assay designed to measure cell surface c-Met receptor may be
conducted to determine if the c-Met antibodies are capable of staining both
human and
cynomolgus monkey c-Met on the cell surface. A secondary anti-mouse-IgG1
antibody
with Alexa Fluor 488 labeling may be used in this assay to detect the cell
surface c-Met
receptors by fluorescence-activated cell surface (F.ACS) analysis.
Briefly described, six-well tissue culture plates may be seeded with MKN45
cells
(human gastric tumor cells that over-express c-Met; Japan Tumor Bank;
#JCRB0254) at
1.5 x 105 cells/well/2 ml in RPM-i640 medium (Life Technologies, Grand Island,
NY;
catalog #11835); 10% FBS (Life Technologies; catalog #10082); 2 mM L-glutamine
(Life Technologies; catalog #25030); 100 U/500 mi., penicillin G and 100
g/500 mI,
streptomycin (Life Technologies; catalog #15140). Plated cells may be
incubated for 24
hours at 37 C, 95% relative humidity, 5% (v/v) CO2. NIH 3T3 cells (ATCC; #CRL-
1658) may be cultured in DMEM (Life Technologies; catalog #11995); 10% calf
serum
(Life Technologies; catalog #30-2030); 2 ruM L-glutamine (Life Technologies;
catalog
#25030); 100 U1500 mL penicillin G and 100 g/500 mL streptomycin (Life
Technologies; catalog #15140).
The cynomolgus monkey c-Met may be introduced into NIH 3T3 cells by
retroviral-mediated gene transfer, and several stable clones over-expressing
cyno c-Met
can be established. NIH 3T3-cyno-c-Met cells may be cultured in six well
tissue culture
plates to 80% confluency. 1 ml of enzyme-free cell dissociation solution (EMU
Millipore, Billerica, Massachusetts; catalog #S-014-B) may be added to each
well, and
left for 5 min. at room temperature to disperse cells. Next, the cells may be
pipetted up

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
29
and down, and collected into centrifuge tubes. The cells may be washed once in
culture
medium followed by one more wash in binding buffer (Dulbecco's phosphate
buffered
saline (DPBS)/I% bovine serum albumin (BSA)/0.01% sodium nide). The c-Met
antibody may be labeled with Alexa Fluor 488 using an Alexa Fluor 488
Monoclonal
Antibody Labeling Kit (Molecular Probes, Eugene, OR; catalog #A-20181)
according to
the manufacturer's instructions. Cell surface c-Met receptors may be stained
with 100 ul
of binding buffer containing 2 lig/m1 of Alexa Fluor 488-labeled c-Met
antibody for 60
min. on ice. Next, the cells may be washed once with binding buffer and
resuspended in
DPBS containing 21.1g/mIpropidium iodide (PI; for staining the dead cells).
The cell
surface c-Met receptors on the live cells may be analyzed by FACS analysis,
and 10,000
events may be acquired for each sample. For data analysis, the mean
fluorescence may
be used to compare anti-c-Met antibody-treated samples with IgG isotype-
treated
controls.
In FACS analysis of c-Met expression on cell surfaces performed essentially as
described in this Example, the OptD11 c-Met antibody stains cell surface c-Met
on
human tumor cells as well as cynomolgus monkey c-Met expressed on NIH 3T3
cells.
Therefore, OptD11 antibodies can be used to stain cell surface c-Met from
fluid of tumor
biopsies, permitting FACS analysis of such samples.
These data indicate that OptD11 c-Met antibody can be used to detect cell
surface
c-Met receptor levels for diagnostic and prognostic purposes, and for
optimizing c-Met
targeted therapies for the treatment of cancer.
Example 4
Human c-Met ECU ELISA using niAb OptD11
The in vitro binding of OptD11 mAb to human c-Met ECD may be determined by
measuring the reactivity of the antibody to human c-Met ECD proteins in an
enzyme-
linked immunosorbent assay (ELISA).
Briefly described, the wells of a 96-well microtiter ELISA plate (Greiner Bio-
One
GmbH; catalog #655081) may be coated with a c-Met capture antibody that
specifically
binds to an epitope within the c-Met ECD a-chain (e.g., SEQ ID NO: 20) andlorn-
chain
(e.g., residues 1-67 or 1-92 of SEQ ID NO: 21). More particularly, the c-Met
capture

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
antibody can be diluted to 2 pg/ml in lx of commercially available coating
buffer (for
example, BioFX catalog # COAT-1000-01 from SurModics 1VD (Eden Prairie, MN)).
Approximately, 110 pL, of the diluted c-Met capture antibody may be added to
each well
of the 96-well microtiter ELISA plate. Then the plate(s) can be covered and
incubated
5 overnight at 4 'C. After the overnight incubation at 4 C, the wells may
be aspirated and
washed two times with 200 pL, wash buffer (BioFX; catalog # WSH-1000-01) using
an
automatic plate-washer. Next, approximately 200 IA, blocking buffer (IX wash
buffer +
2% BSA) (Sigma-Aldrich Co., St. Louis, MO, catalog # A7979) may be added to
each
well of the ELISA plate and incubated 1 hour at room temperature. Next, the
wells of the
10 plates may be washed two times with approximately 200 L wash buffer
using an
automatic plate-washer. One hundred microliters (ML) of serially diluted (in
an
acceptable diluent, e.g., blocking buffer) ECD of human c-Met expressed as a
Fc fusion
protein with a flag- and His-tag (His-tag) at the C-terminus of the Fc (as
shown in SEQ
ID NO: 17, for example) may be added to the ELISA plate(s) and incubated for 2
hours
15 with shaking at room temperature. The serial dilutions may start at 1280
pg/mL with 2-
fold dilution down to 20 pg/mL, for example. Next, 100 ML of 0.5 pg/mL
biotinylated
OptD11 c-Met antibody (in an acceptable diluent, e.g., blocking buffer) may be
added as
the c-Met detection antibody and incubated 2 hours with shaking at room
temperature.
The plates may then be aspirated and washed 4 times with wash buffer using
automatic
20 plate-washer. Next, 100 ML of 83 ng/mL peroxidase conjugated
streptavidin (Jackson
Immunoresearch, West Grove, PA; catalog # 016-030-084) in diluent may be added
to all
the wells and the plates may be incubated two hours with shaking at room
temperature.
Next the wells of the plates may be aspirated and washed 6 times with wash
buffer using
automatic plate-washer. After the plates are washed, 100 pt of TMB (BioFX
catalog #
25 TMBW-1000-01) may be added to each well and the plates may be incubated
for 10
minutes at room temperature. To stop the reaction, 100 pL, of stop solution
may be added
to each well (BioFX; catalog # LSTP-1000-01). The colorimetric signals may be
developed and read using a plate reader at 450 nm with 570 nm correction.
The monoclonal antibody OptD11 was tested for binding to recombinant human
30 c-Met ECD protein fused to Fe in an ELISA essentially as described
above. More
specifically, using the anti-c-Met antibody C8-H241 (human IgG4 subtype) as c-
Met

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
31
capture antibody and biotinylated mAb OptD11 as the c-Met detection antibody
reagent,
mAb OptD11 provides high sensitivity (detects c-Met ECD as low as 20 pg/ml) as
well as
a wide range of c-Met ECD detection (i.e., 20 pg/m1 to 1280 pg/m1) (see Figure
2). The
results of the ELISA experiment indicate that OptD11 binds to a recombinant
human
c-Met ECD fusion protein and may be useful in in vitro immunochemical methods
including, but not limited to, ELISA, for detecting the ECD of c-Met receptor
for
diagnostic, prognostic and predictive purposes.
Example 5
mAb OptD11 and mAb C8-H241 Simultaneously Bind ECD of Human c-Met
To determine if an anti-c-Met antibody of the invention (such as OptD11) can
bind the ECD of human c-Met at the same time as an anti-c-Met therapeutic
antibody
(such as, e.g., C8-H241, disclosed in WO 2010/059654 and/or U.S. Patent No.
8,217,148), a binding experiment may be performed using a surface plasmon
resonance
biosensor such as a BIAcore 2000, BlAcoree 3000, or a BIAcore T100 (GE
Health
Care, Piscataway, NJ) according to methods known in the art. Except as noted,
all
reagents and materials may be purchased from BIAcore . All reagents and
materials may
be purchased from BIAcoree unless otherwise noted. All measurements may be
performed at 25 C using HBS-EP (150 mM sodium chloride, 3 mM EDTA, 0.005 %
(w/v) surfactant P-20, and 10 mM HEPES, pH 7.4) as both a running and sample
buffer.
Mouse IgG1 anti-polyhistidine antibody (R&D systems, Mab050) may be
immobilized
on flow cells 1 to 4 of a CM5 sensor chip at a level of 7000 response units
(Rus) using an
amine coupling kit. Next, human c-Met-ECD-Fc-Flis may be injected to flow cell
2 then
an anti-c-Met antibody (500 riM), such as mAb C8-H241, may be injected through
flow
cell 1 and 2 (50 L/min for 5 minutes). By the end of injection, binding may
be saturated
and a binding response unit may be used to calculate binding stoichiometry.
In experiments performed essentially as described in this Example 5, each c-
Met-
ECD-Fc-Flis dimer binds 0.9 C8-H241 (human IgG4 subtype) anti-c-Met
antibodies.
Upon subsequent injection of mAb OptD11 (mouse IgG1 subtype) additional
binding to
c-Met-ECD-Fe-Flis was observed (see Figure 3). The stoichiometry of the
additional

CA 02843771 2014-01-30
WO 2013/043452
PCT/US2012/055057
32
binding was also approximately 0.9, indicating that anti-c-Met mAb Optll11
(mouse
IgG I subtype) also filly binds c-Met-ECD-Fc-Flis that is bound by anti-C-met
mAb C8-
H241 (human IgG4 subtype).
In order to determine whether simultaneous binding of anti-c-Met mAbs C8-H241
(human IgG4 subtype) and OptD I I (mouse IgG1 subtype) to human c-Met-ECD-Fc-
Flis
is independent of binding order, the chip surface was regenerated to remove c-
Met-ECD-
Fc-Flis and both c-Met antibodies before repeating the binding cycle. An
additional cycle
of binding was repeated as described above, however, the binding order of the
antibodies
was reversed (i.e., mAb OptD11 before mAb C8-H241). Similar results were
obtained
(see Figure 3).
These data show that mAb OptD11 (mouse IgG I subtype) and mAb C8-H241
(human IgG4 subtype) can simultaneously bind human c-Met-ECD-Fc-Flis.
Furthermore, the data strongly suggests that the epi topes of these two anti-c-
Met
antibodies differ.

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Inactive: Dead - No reply to s.30(2) Rules requisition 2017-11-07
Application Not Reinstated by Deadline 2017-11-07
Deemed Abandoned - Failure to Respond to Maintenance Fee Notice 2017-09-13
Inactive: Abandoned - No reply to s.30(2) Rules requisition 2016-11-07
Inactive: S.30(2) Rules - Examiner requisition 2016-05-06
Inactive: Report - No QC 2016-05-05
Amendment Received - Voluntary Amendment 2015-10-26
Inactive: S.30(2) Rules - Examiner requisition 2015-04-24
Inactive: Report - No QC 2015-04-23
Inactive: Sequence listing - Amendment 2014-12-17
BSL Verified - Defect(s) 2014-12-17
BSL Verified - No Defects 2014-12-17
Inactive: Compliance - PCT: Resp. Rec'd 2014-12-17
Inactive: Incomplete PCT application letter 2014-12-09
Inactive: Cover page published 2014-03-12
Inactive: IPC removed 2014-03-04
Application Received - PCT 2014-03-04
Inactive: First IPC assigned 2014-03-04
Inactive: IPC assigned 2014-03-04
Inactive: IPC assigned 2014-03-04
Inactive: IPC assigned 2014-03-04
Inactive: IPC assigned 2014-03-04
Inactive: First IPC assigned 2014-03-04
Inactive: IPC removed 2014-03-04
Inactive: IPC assigned 2014-03-04
Inactive: IPC assigned 2014-03-04
Inactive: Acknowledgment of national entry - RFE 2014-03-04
Letter Sent 2014-03-04
National Entry Requirements Determined Compliant 2014-01-30
Request for Examination Requirements Determined Compliant 2014-01-30
Inactive: Sequence listing - Refused 2014-01-30
BSL Verified - Defect(s) 2014-01-30
Inactive: Sequence listing - Amendment 2014-01-30
Amendment Received - Voluntary Amendment 2014-01-30
All Requirements for Examination Determined Compliant 2014-01-30
Application Published (Open to Public Inspection) 2013-03-28

Abandonment History

Abandonment Date Reason Reinstatement Date
2017-09-13

Maintenance Fee

The last payment was received on 2016-08-17

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Fee History

Fee Type Anniversary Year Due Date Paid Date
Basic national fee - standard 2014-01-30
Request for examination - standard 2014-01-30
MF (application, 2nd anniv.) - standard 02 2014-09-15 2014-08-28
2014-12-17
MF (application, 3rd anniv.) - standard 03 2015-09-14 2015-08-18
MF (application, 4th anniv.) - standard 04 2016-09-13 2016-08-17
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
ELI LILLY AND COMPANY
Past Owners on Record
JIRONG LU
JULIAN DAVIES
LING LIU
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column (Temporarily unavailable). To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2014-01-29 32 2,398
Representative drawing 2014-01-29 1 22
Drawings 2014-01-29 3 85
Claims 2014-01-29 9 516
Abstract 2014-01-29 2 76
Claims 2014-01-30 6 222
Cover Page 2014-03-11 1 45
Description 2015-10-25 32 2,291
Claims 2015-10-25 6 232
Acknowledgement of Request for Examination 2014-03-03 1 177
Notice of National Entry 2014-03-03 1 203
Reminder of maintenance fee due 2014-05-13 1 111
Courtesy - Abandonment Letter (Maintenance Fee) 2017-10-24 1 174
Courtesy - Abandonment Letter (R30(2)) 2016-12-18 1 164
PCT 2014-01-29 6 189
Correspondence 2014-12-08 2 45
Correspondence 2014-12-16 2 56
Amendment / response to report 2015-10-25 13 523
Examiner Requisition 2016-05-05 3 195

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :